Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Pharmacol Sin ; 41(11): 1465-1475, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32341465

RESUMO

Peptide transporter 1 (PepT1), highly expressed on the apical membrane of enterocytes, is involved in energy balance and mediates intestinal absorption of peptidomimetic drugs. In this study, we investigated whether and how diabetes affected the function and expression of intestinal PepT1. Diabetes was induced in rats by combination of high-fat diet and low dose streptozocin injection. Pharmacokinetics study demonstrated that diabetes significantly decreased plasma exposures of cephalexin and acyclovir following oral administration of cephalexin and valacyclovir, respectively. Single-pass intestinal perfusion analysis showed that diabetes remarkably decreased cephalexin absorption, which was associated with decreased expression of intestinal PepT1 protein. We assessed the levels of bile acids in intestine of diabetic rats, and found that diabetic rats exhibited significantly higher levels of chenodeoxycholic acid (CDCA), cholic acid (CA) and glycocholic acid (GCA), and lower levels of lithocholic acid (LCA) and hyodeoxycholic acid (HDCA) than control rats; intestinal deoxycholic acid (DCA) levels were unaltered. In Caco-2 cells, the 6 bile acids remarkably decreased expression of PepT1 protein with CDCA causing the strongest inhibition, whereas TNF-α, LPS and insulin little affected expression of PepT1 protein; short-chain fatty acids induced rather than decreased expression of PepT1 protein. Farnesoid X receptor (FXR) inhibitor glycine-ß-muricholic acid or FXR knockdown reversed the downregulation of PepT1 expression by CDCA and GW4064 (another FXR agonist). In diabetic rats, the expression of intestinal FXR protein was markedly increased. Oral administration of CDCA (90, 180 mg·kg-1·d-1, for 3 weeks) dose-dependently decreased the expression and function of intestinal PepT1 in rats. In conclusion, diabetes impairs the expression and function of intestinal PepT1 partly via CDCA-mediated FXR activation.


Assuntos
Ácidos Cólicos/farmacologia , Diabetes Mellitus Experimental/fisiopatologia , Regulação para Baixo/fisiologia , Transportador 1 de Peptídeos/metabolismo , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Animais , Células CACO-2 , Cefalexina/metabolismo , Cefalexina/farmacocinética , Ácidos Cólicos/metabolismo , Humanos , Jejuno/metabolismo , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Valaciclovir/metabolismo , Valaciclovir/farmacocinética
2.
Mol Pharm ; 17(5): 1706-1714, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32196348

RESUMO

Prodrug discovery and development in the pharmaceutical industry have been hampered by a lack of knowledge of prodrug activation pathways. Such knowledge would minimize the risks of prodrug failure by enabling proper selection of preclinical animal models, prediction of pharmacogenomic variability, and identification of drug-drug interactions. Technologies for annotation of activating enzymes have not kept pace with the growing need. Activity-based protein profiling (ABPP) has matured considerably in recent decades, leading to widespread use in the pharmaceutical industry. Here, we report the extension of competitive ABPP (cABPP) to prodrug-activating enzyme identification in stable isotope-labeled cell lysates using a modified fluorophosphonate probe. Focusing on the antiviral ester prodrug valacyclovir (VACV), we identified serine hydrolase RBBP9 as an activating enzyme in Caco-2 cells via shotgun proteomics, validating the activity via the selective inhibitor emetine (EME). Kinetic characterization of RBBP9 revealed a catalytic efficiency (kcat·KM-1 = 104 mM-1·s-1) comparable to that of BPHL, the only known VACV-activating enzyme prior to this work. EME incubation in wild-type and Bphl-knockout jejunum and liver lysates demonstrated the near-exclusivity of VACV activation by RBBP9 in the intestine. Additionally, these studies showed that RBBP9 and BPHL are the two major and coequal VACV-activating enzymes in the liver. Single-pass intestinal perfusions of VACV ± EME in mice showed EME coperfusion significantly inhibited the intestinal activation of VACV, implying the in vivo relevance of RBBP9-mediated VACV activation. We envision that others might use the cABPP approach in the future for global, rapid, and efficient discovery of prodrug-activating enzymes.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Proteômica/métodos , Valaciclovir/metabolismo , Ativação Metabólica , Animais , Células CACO-2 , Proteínas de Ciclo Celular/antagonistas & inibidores , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/antagonistas & inibidores , Pró-Fármacos/metabolismo
3.
Biochem Pharmacol ; 156: 147-156, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30121252

RESUMO

Biphenyl hydrolase-like protein (BPHL) is a novel human serine hydrolase that was originally cloned from a breast carcinoma cDNA library and shown to convert valacyclovir to acyclovir and valganciclovir to ganciclovir. However, the exclusivity of this process has not been determined and, indeed, it is possible that a number of esterases/proteases may mediate the hydrolysis of valacyclovir and similar prodrugs. The objectives of the present study were to evaluate the in situ intestinal permeability and stability of valacyclovir in wildtype (WT) and Bphl knockout (KO) mice, as well as the in vivo oral absorption and intravenous disposition of valacyclovir and acyclovir in the two mouse genotypes. We found that Bphl knockout mice had no obvious phenotype and that Bphl ablation did not alter the jejunal permeability of valacyclovir during in situ perfusions (i.e., 0.54 × 10-4 in WT vs. 0.53 × 10-4 cm/s in KO). Whereas no meaningful changes occurred between genotypes in the gene expression of proton-coupled oligopeptide transporters (i.e., PepT1, PepT2, PhT1, PhT2), enzymatic upregulation of Cyp3a11, Cyp3a16, Abhd14a and Abhd14b was observed in some tissues of Bphl knockout mice. Most importantly, we found that valacyclovir was rapidly and efficiently hydrolyzed to acyclovir in the absence of BPHL, and that hydrolysis was more extensive after the oral vs. intravenous route of administration (for both genotypes). Taken as a whole, BPHL is not obligatory for the conversion of valacyclovir to acyclovir either presystemically or systemically.


Assuntos
Antivirais/farmacocinética , Hidrolases de Éster Carboxílico/metabolismo , Valaciclovir/farmacocinética , Administração Oral , Animais , Antivirais/metabolismo , Área Sob a Curva , Hidrolases de Éster Carboxílico/genética , Meia-Vida , Injeções Intravenosas , Camundongos , Camundongos Knockout , Valaciclovir/administração & dosagem , Valaciclovir/metabolismo
4.
Biochem Pharmacol ; 155: 1-7, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29935147

RESUMO

The purpose of this work was to evaluate the intestinal permeability, oral absorption and disposition of the ester prodrug valacyclovir in wildtype mice and a huPepT1 transgenic mouse model. PepT1 (SLC15A1) is a transporter apically expressed along the lumen of the gastrointestinal tract and is responsible for the absorption of di-/tripeptides, ACE inhibitors, ß-lactam antibiotics and numerous prodrugs. Unfortunately, PepT1-mediated substrates that have been extensively studied were shown to exhibit species-dependent absorption and pharmacokinetics. Accordingly, in situ intestinal perfusion studies were conducted and valacyclovir uptake was shown to have a 30-fold lower Km and 100-fold lower Vmax in huPepT1 compared to wildtype mice. Moreover, inhibition studies demonstrated that the huPepT1 transporter alone was responsible for valacyclovir uptake, and segment-dependent studies reported significant reductions in permeability along the length of small intestine in huPepT1 mice. Subsequent oral administration studies revealed that the in vivo rate and extent of valacyclovir absorption were lower in huPepT1 mice, as indicated by 3-fold lower Cmax and 3-fold higher Tmax values, and an AUC0-180 that was 80% of that observed in wildtype mice. However, no significant changes in drug disposition were observed between genotypes after intravenous bolus administration of acyclovir. Lastly, mass balance studies established that the bioavailability of acyclovir, after oral dosing of valacyclovir, was 77.5% in wildtype mice and 52.8% in huPepT1 mice, which corroborated values of 51.3% in clinical studies. Thus, it appears the huPepT1 transgenic mice may be a better model to study prodrug absorption and disposition in humans than wildtype mice.


Assuntos
Antivirais/metabolismo , Absorção Intestinal/fisiologia , Mucosa Bucal/metabolismo , Transportador 1 de Peptídeos/biossíntese , Pró-Fármacos/metabolismo , Valaciclovir/metabolismo , Administração Oral , Animais , Antivirais/administração & dosagem , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Absorção Intestinal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mucosa Bucal/efeitos dos fármacos , Transportador 1 de Peptídeos/genética , Pró-Fármacos/administração & dosagem , Valaciclovir/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...